Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Angiogenesis ; 22(4): 491-505, 2019 11.
Article in English | MEDLINE | ID: mdl-31256320

ABSTRACT

Neovascularization of the inner retinal space is a major cause of vision loss. In retinal angiomatous proliferation (RAP) syndrome, newly formed vessels originate from the retinal plexus and invade the inner retinal space. However, the molecular pathways preventing subretinal vascularization remain largely unknown. In most murine models of RAP, pathological neovascularization occurs concomitantly with the development of the retinal vasculature. Here, we demonstrate that disturbing the sequence of morphogenetic events that shape the three-layered retinal vascular network leads to subretinal vascularization. Sprouts emerging from the perivenous region after the first postnatal week extended toward the retinal space where they merged into the deep layer. The small GTPase Rac1 was required for the formation of these vascular extensions and the vascular inner plexus is formed coaxially to the overarching veins. The adhesion receptor Adgrf5 was highly expressed in the endothelium of the central nervous system, where it regulates blood-brain barrier formation. The vascular superficial plexus of Adgrf5 mutant mouse retinae exhibited an increased vascular density in the perivenous areas with increased projections toward the inner plexus where they subsequently created hyper-dense endothelial cells (EC) clusters. Disturbing the perivenous pool of EC thus significantly altered the inner plexus formation. These abnormalities culminated in transient vascular protrusions in the inner retinal space. Taken together, these results reveal a previously unobserved vascular morphogenetic defect in Adgrf5 knockout mice, implicating a role for ADGRF5 in the initiation of subretinal vascularization. Our findings also illustrate how vein-derived EC shape the inner retinal layer formation and could control the appearance of angiomatous malformations.


Subject(s)
Endothelium, Vascular/metabolism , Receptors, G-Protein-Coupled/metabolism , Retina/metabolism , Retinal Neovascularization/metabolism , Animals , Endothelium, Vascular/pathology , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Retina/pathology , Retinal Neovascularization/pathology
2.
Nat Commun ; 10(1): 1448, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30923344

ABSTRACT

The original version of this Article omitted the following from the Acknowledgements: 'This project was supported by CRC128/Project A03 of the Deutsche Forschungsgemeinschaft (DFG).'This has not been corrected in either the PDF or HTML versions.

3.
Acta Neuropathol Commun ; 5(1): 42, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28578681

ABSTRACT

Bioactive lipids contribute to the pathophysiology of multiple sclerosis. Here, we show that lysophosphatidic acids (LPAs) are dysregulated in multiple sclerosis (MS) and are functionally relevant in this disease. LPAs and autotaxin, the major enzyme producing extracellular LPAs, were analyzed in serum and cerebrospinal fluid in a cross-sectional population of MS patients and were compared with respective data from mice in the experimental autoimmune encephalomyelitis (EAE) model, spontaneous EAE in TCR1640 mice, and EAE in Lpar2 -/- mice. Serum LPAs were reduced in MS and EAE whereas spinal cord LPAs in TCR1640 mice increased during the 'symptom-free' intervals, i.e. on resolution of inflammation during recovery hence possibly pointing to positive effects of brain LPAs during remyelination as suggested in previous studies. Peripheral LPAs mildly re-raised during relapses but further dropped in refractory relapses. The peripheral loss led to a redistribution of immune cells from the spleen to the spinal cord, suggesting defects of lymphocyte homing. In support, LPAR2 positive T-cells were reduced in EAE and the disease was intensified in Lpar2 deficient mice. Further, treatment with an LPAR2 agonist reduced clinical signs of relapsing-remitting EAE suggesting that the LPAR2 agonist partially compensated the endogenous loss of LPAs and implicating LPA signaling as a novel treatment approach. Graphical summary of lysophosphatidic signaling in multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Lysophospholipids/metabolism , Multiple Sclerosis/metabolism , Adolescent , Adult , Animals , Biomarkers/metabolism , Cohort Studies , Cross-Sectional Studies , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Immunologic Factors/pharmacology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Young Adult
4.
Nat Commun ; 8: 15700, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28621310

ABSTRACT

G-protein-coupled receptor (GPCR) expression is extensively studied in bulk cDNA, but heterogeneity and functional patterning of GPCR expression in individual vascular cells is poorly understood. Here, we perform a microfluidic-based single-cell GPCR expression analysis in primary smooth muscle cells (SMC) and endothelial cells (EC). GPCR expression is highly heterogeneous in all cell types, which is confirmed in reporter mice, on the protein level and in human cells. Inflammatory activation in murine models of sepsis or atherosclerosis results in characteristic changes in the GPCR repertoire, and we identify functionally relevant subgroups of cells that are characterized by specific GPCR patterns. We further show that dedifferentiating SMC upregulate GPCRs such as Gpr39, Gprc5b, Gprc5c or Gpr124, and that selective targeting of Gprc5b modulates their differentiation state. Taken together, single-cell profiling identifies receptors expressed on pathologically relevant subpopulations and provides a basis for the development of new therapeutic strategies in vascular diseases.


Subject(s)
Cell Differentiation , Inflammation , Myocytes, Smooth Muscle/cytology , Receptors, G-Protein-Coupled/metabolism , Animals , Aorta/pathology , Atherosclerosis/metabolism , Cluster Analysis , Exons , Green Fluorescent Proteins/metabolism , Humans , Ligands , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Sepsis/metabolism , Sepsis/physiopathology , Single-Cell Analysis , Tissue Array Analysis
5.
J Thromb Haemost ; 13(11): 2102-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26334261

ABSTRACT

BACKGROUND: RhoA is an important regulator of platelet responses downstream of Gα13 , yet we still know little about its regulation in platelets. Leukemia-associated Rho guanine-nucleotide exchange factor (GEF [LARG]), a RhoA GEF, is highly expressed in platelets and may constitute a major upstream activator of RhoA. To this end, it is important to determine the role of LARG in platelet function and thrombosis. METHODS AND RESULTS: Using a platelet-specific gene knockout, we show that the absence of LARG results in a marked reduction in aggregation and dense-granule secretion in response to the thromboxane mimetic U46619 and proteinase-activated receptor 4-activating peptide, AYPGKF, but not to adenosine diphosphate. In a ferric chloride thrombosis model in vivo, this translated into a defect, under mild injury conditions. Importantly, agonist-induced RhoA activation was not affected by the absence of LARG, although basal activity was reduced, suggesting that LARG may play a housekeeper role in regulating constitutive RhoA activity. CONCLUSIONS: LARG plays an important role in platelet function and thrombosis in vivo. However, although LARG may have a role in regulating the resting activation state of RhoA, its role in regulating platelet function may principally be through RhoA-independent pathways, possibly through other Rho family members.


Subject(s)
Blood Platelets/metabolism , Platelet Activation/physiology , Rho Guanine Nucleotide Exchange Factors/physiology , Thrombosis/blood , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/drug effects , Cell Degranulation/drug effects , Chlorides/toxicity , Crosses, Genetic , Ferric Compounds/toxicity , Gene Knockout Techniques , Mice , Mice, Knockout , Oligopeptides/pharmacology , Organ Specificity , Platelet Aggregation , Rho Guanine Nucleotide Exchange Factors/blood , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/genetics , Thrombosis/chemically induced
6.
Neuroscience ; 290: 126-37, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25637492

ABSTRACT

G-protein-coupled receptor 41 (GPR41) also called free fatty acid receptor 3 (FFAR3) is a Gαi-coupled receptor activated by short-chain fatty acids (SCFAs) mainly produced from dietary complex carbohydrate fibers in the large intestine as products of fermentation by microbiota. FFAR3 is expressed in enteroendocrine cells, but has recently also been shown to be present in sympathetic neurons of the superior cervical ganglion. The aim of this study was to investigate whether the FFAR3 is present in other autonomic and sensory ganglia possibly influencing gut physiology. Cryostat sections were cut of autonomic and sensory ganglia of a transgenic reporter mouse expressing the monomeric red fluorescent protein (mRFP) gene under the control of the FFAR3 promoter. Control for specific expression was also done by immunohistochemistry with an antibody against the reporter protein. mRFP expression was as expected found not only in neurons of the superior cervical ganglion, but also in sympathetic ganglia of the thoracic and lumbar sympathetic trunk. Further, neurons in prevertebral ganglia expressed the mRFP reporter. FFAR3-mRFP-expressing neurons were also present in both autonomic and sensory ganglia such as the vagal ganglion, the spinal dorsal root ganglion and the trigeminal ganglion. No expression was observed in the brain or spinal cord. By use of radioactive-labeled antisense DNA probes, mRNA encoding the FFAR3 was found to be present in cells of the same ganglia. Further, the expression of the FFAR3 in the ganglia of the transgenic mice was confirmed by immunohistochemistry using an antibody directed against the receptor protein, and double labeling colocalized mRFP and the FFAR3-protein in the same neurons. Finally, quantitative real-time polymerase chain reaction (qRT-PCR) on extracts from the ganglia supported the presence mRNA encoding the FFAR3 in most of the investigated tissues. These data indicate that FFAR3 is expressed on postganglionic sympathetic and sensory neurons in both the autonomic and somatic peripheral nervous system and that SCFAs act not only through the enteroendocrine system but also directly by modifying physiological reflexes integrating the peripheral nervous system and the gastro-intestinal tract.


Subject(s)
Ganglia, Spinal/metabolism , Ganglia, Sympathetic/metabolism , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Trigeminal Ganglion/metabolism , Animals , Autoradiography , Brain/metabolism , Immunohistochemistry , In Situ Hybridization , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Photomicrography , Promoter Regions, Genetic , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/genetics , Spinal Cord/metabolism , Red Fluorescent Protein
7.
Clin Exp Immunol ; 177(1): 179-89, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24628494

ABSTRACT

In different bioassays, functional antibodies reacting with the human muscarinic acetylcholine receptor M3(mAchR3) have been detected in sera from patients with Sjögren's syndrome (SS), and there is strong evidence that those antibodies may have pathogenetic relevance. However, depending on the method of detection, their prevalence varied. Furthermore, those bioassays are difficult to standardize. We report on the development and optimization of a novel test system based on a luminometric method to determine downstream signalling of mAchR3 which produces specific and reproducible results. Chinese hamster ovarian (CHO) cells were transfected with plasmids encoding mAchR3 and a green fluorescence protein (GFP)/aequorin fusion protein. Incubation of cells with carbachol resulted in an increase in intracellular [Ca(2+)], which was detected by measuring light emission with a luminometer, and the effect of incubation with patients' immunoglobulins (Ig) was evaluated. Optimal cell density, Ig preparation and time of incubation with patients' sera were determined. Sera from patients with primary Sjögren's syndrome (pSS; n = 40), systemic sclerosis (SSc; n = 47), myasthenia gravis (MG; n = 133) and 50 blood donors were analysed. Optimal assay conditions were obtained with a cell density of 100 000 cells/ml, isolation of Ig by ammonium sulphate precipitation and short-term incubation. Based on this highly reliable assay, 50% of the pSS patients had antibodies which inhibited carbachol-induced activation of mAchR3; none of the SSc patients, 6% of the patients with MG and 12% of the blood donors had antibodies which reacted with the mAchR3. This method facilitates the determination of functional anti-mAchR3 antibodies in patients' sera, confirmed their high prevalence in pSS patients and may, therefore, help to analyse their pathogenetic and clinical relevance in more detail.


Subject(s)
Autoantibodies/blood , Luminescent Measurements , Myasthenia Gravis/diagnosis , Receptor, Muscarinic M3/immunology , Scleroderma, Systemic/diagnosis , Serologic Tests/methods , Sjogren's Syndrome/diagnosis , Adolescent , Adult , Aged , Aged, 80 and over , Animals , CHO Cells , Calcium Signaling , Cricetinae , Cricetulus , Humans , Middle Aged , Myasthenia Gravis/immunology , Prevalence , Reproducibility of Results , Scleroderma, Systemic/immunology , Sensitivity and Specificity , Sjogren's Syndrome/epidemiology , Sjogren's Syndrome/immunology , Young Adult
8.
Biochim Biophys Acta ; 1832(8): 1149-58, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23579073

ABSTRACT

Elevated levels of homocysteine produce detrimental effects in humans but its role in preterm birth is not known. Here we used a mouse model of hyperhomocysteinemia to examine the relevance of homocysteine to preterm birth. The mouse carries a heterozygous deletion of cystathionine ß-synthase (Cbs(+/-)). Gestational period was monitored in wild type and Cbs(+/-) female mice. Mouse uterine and placental tissues, human primary trophoblast cells, and human myometrial and placental cell lines were used to determine the influence of homocysteine on expression of specific genes in vitro. The activity of BKCa channel in the myometrial cell line was monitored using the patch-clamp technique. We found that hyperhomocysteinemia had detrimental effects on pregnancy and induced preterm birth in mice. Homocysteine increased the expression of oxytocin receptor and Cox-2 as well as PGE2 production in uterus and placenta, and initiated premature uterine contraction. A Cox-2 inhibitor reversed these effects. Gpr109a, a receptor for niacin, induced Cox-2 in uterus. Homocysteine upregulated GPR109A and suppressed BKCa channel activity in human myometrial cells. Deletion of Gpr109a in Cbs(+/-) mice reversed premature birth. We conclude that hyperhomocysteinemia causes preterm birth in mice through upregulation of the Gpr109a/Cox-2/PGE2 axis and that pharmacological blockade of Gpr109a may have potential in prevention of preterm birth.


Subject(s)
Homocysteine/blood , Hyperhomocysteinemia/physiopathology , Pregnancy Complications/blood , Premature Birth/blood , Animals , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Dinoprostone/genetics , Dinoprostone/metabolism , Female , Homocysteine/genetics , Homocysteine/metabolism , Humans , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Mice , Mice, Inbred C57BL , Muscle Contraction , Myometrium/metabolism , Myometrium/physiopathology , Placenta/metabolism , Placenta/physiopathology , Pregnancy , Pregnancy Complications/genetics , Pregnancy Complications/physiopathology , Premature Birth/genetics , Premature Birth/physiopathology , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Trophoblasts/metabolism , Up-Regulation , Uterus/metabolism , Uterus/pathology , Uterus/physiopathology
9.
Mol Cell Biol ; 30(3): 764-80, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19948886

ABSTRACT

Semaphorins and their receptors, plexins, are emerging as key regulators of various aspects of neural and nonneural development. Semaphorin 4D (Sema4D) and B-type plexins demonstrate distinct expression patterns over critical time windows during the development of the murine neocortex. Here, analysis of mice genetically lacking plexin-B1 or plexin-B2 revealed the significance of Sema4D-plexin-B signaling in cortical development. Deficiency of plexin-B2 resulted in abnormal cortical layering and defective migration and differentiation of several subtypes of cortical neurons, including Cajal-Retzius cells, GABAergic interneurons, and principal cells in vivo. In contrast, a lack of plexin-B1 did not impact on cortical development in vivo. In various ex vivo assays on embryonic forebrain, Sema4D enhanced the radial and tangential migration of developing neurons in a plexin-B2-dependent manner. These results suggest that Sema4D-plexin-B2 interactions regulate mechanisms underlying cell specification, differentiation, and migration during corticogenesis.


Subject(s)
Neocortex/embryology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , Animals , Cell Movement/genetics , Cell Movement/physiology , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Mutation/genetics , Mutation/physiology , Neocortex/cytology , Neocortex/metabolism , Nerve Tissue Proteins/genetics , Neurons/cytology , Receptors, Cell Surface/genetics , Sequence Deletion/genetics , Sequence Deletion/physiology
10.
Br J Pharmacol ; 153 Suppl 1: S68-75, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18037924

ABSTRACT

Nicotinic acid has been used for decades to treat dyslipidaemic states. In particular its ability to raise the plasma HDL cholesterol concentration has led to an increased interest in its pharmacological potential. The clinical use of nicotinic acid is somewhat limited due to several harmless but unpleasant side effects, most notably a cutaneous flushing phenomenon. With the recent discovery of a nicotinic acid receptor, it has become possible to better understand the mechanisms underlying the metabolic and vascular effects of nicotinic acid. Based on these new insights into the action of nicotinic acid, novel strategies are currently under development to maximize the pharmacological potential of this drug. The generation of both flush-reducing co-medications of nicotinic acid and novel drugs targeting the nicotinic acid receptor will provide future therapeutic options for the treatment of dyslipidaemic disorders.


Subject(s)
Dyslipidemias/drug therapy , Hypolipidemic Agents/therapeutic use , Niacin/pharmacology , Animals , Dyslipidemias/blood , Flushing/chemically induced , Humans , Hypolipidemic Agents/adverse effects , Hypolipidemic Agents/chemistry , Hypolipidemic Agents/pharmacokinetics , Lipid Metabolism/drug effects , Niacin/adverse effects , Niacin/chemistry , Niacin/pharmacokinetics , Niacin/therapeutic use , Receptors, Nicotinic/drug effects
11.
Handb Exp Pharmacol ; (178): 491-509, 2007.
Article in English | MEDLINE | ID: mdl-17203668

ABSTRACT

The G-protein-coupled receptor signaling system, consisting of a huge variety of receptors as well as of many G-proteins and effectors, operates in every cell and is involved in many physiological and pathological processes. The versatility of this system and the involvement of specific components makes G-protein-coupled receptors and their signaling pathways ideal targets for pharmacological interventions. Classical mouse knockout models have often provided important preliminary insights into the biological roles of individual receptors and signaling pathways and they are routinely used in the process of target validation. The recent development of efficient conditional mutagenesis techniques now allows a much more detailed analysis of G-protein-mediated signaling transduction processes. This review summarizes some of the areas in which progress has recently been made by applying conditional mutagenesis of genes coding for G-proteins and G-protein-coupled receptors.


Subject(s)
GTP-Binding Proteins/metabolism , Mutagenesis , Receptors, G-Protein-Coupled/metabolism , Animals , Blood Vessels/embryology , Neural Crest/embryology , Platelet Activation/physiology
12.
J Thromb Haemost ; 3(10): 2296-306, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16194206

ABSTRACT

The dynamics of the actin cytoskeleton, largely controlled by the Rho family of small GTPases (Rho, Rac and Cdc42), is critical for the regulation of platelet responses such as shape change, adhesion, spreading and aggregation. Here, we investigated the role of adenosine diphosphate (ADP), a major co-activator of platelets, on the activation of Rac. ADP rapidly activated Rac in a dose-dependent manner and independently of GPIIb/IIIa and phosphoinositide 3-kinase. ADP alone, used as a primary agonist, activated Rac and its effector PAK via its P2Y1 receptor, through a G(q)-dependent pathway and independently of P2Y12. The P2Y12 receptor appeared unable to activate the GTPase per se as also observed for the adenosine triphosphate receptor P2X1. Conversely, secreted ADP strongly potentiated Rac activation induced by FcgammaRIIa clustering or TRAP via its P2Y12 receptor, the target of antithrombotic thienopyridines. Stimulation of the alpha(2A)-adrenergic receptor/G(z) pathway by epinephrine was able to replace the P2Y12/G(i)-mediated pathway to amplify Rac activation by FcgammaRIIa or by the thrombin receptor PAR-1. This co-activation appeared necessary to reach a full stimulation of Rac as well as PAK activation and actin polymerization and was blocked by a G-protein betagamma subunits scavenger peptide.


Subject(s)
Blood Platelets/metabolism , Membrane Proteins/physiology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Purinergic P2/physiology , Actins/metabolism , Adenosine Diphosphate/pharmacology , Animals , Dose-Response Relationship, Drug , GTP Phosphohydrolases/metabolism , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Receptors, Purinergic P2Y1 , Receptors, Purinergic P2Y12 , p21-Activated Kinases
13.
Mol Cell Biol ; 25(5): 1942-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15713647

ABSTRACT

Heterotrimeric G proteins of the Gq/11 family transduce signals from a variety of neurotransmitter and hormone receptors and have therefore been implicated in various functions of the nervous system. Using the Cre/loxP system, we generated mice which lack the genes coding for the alpha subunits of the two main members of the Gq/11 family, gnaq and gna11, selectively in neuronal and glial precursor cells. Mice with defective gnaq and gna11 genes were morphologically normal, but they died shortly after birth. Mice carrying a single gna11 allele survived the early postnatal period but died within 3 to 6 weeks as anorectic dwarfs. In these mice, postnatal proliferation of pituitary somatotroph cells was strongly impaired, and plasma growth hormone (GH) levels were reduced to 15%. Hypothalamic levels of GH-releasing hormone (GHRH), an important stimulator of somatotroph proliferation, were strongly decreased, and exogenous administration of GHRH restored normal proliferation. The hypothalamic effects of ghrelin, a regulator of GHRH production and food intake, were reduced in these mice, suggesting that an impairment of ghrelin receptor signaling might contribute to GHRH deficiency and abnormal eating behavior. Taken together, our findings show that Gq/11 signaling is required for normal hypothalamic function and that impairment of this signaling pathway causes somatotroph hypoplasia, dwarfism, and anorexia.


Subject(s)
Dwarfism, Pituitary/etiology , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Growth Hormone-Releasing Hormone/metabolism , Hypothalamus/metabolism , Pituitary Gland/pathology , Alleles , Animals , Cell Proliferation/drug effects , Dwarfism, Pituitary/metabolism , Eating/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/analysis , Ghrelin , Growth Hormone/analysis , Growth Hormone/metabolism , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/pharmacology , Hypothalamus/chemistry , Hypothalamus/drug effects , Mice , Mice, Knockout , Mutation/genetics , Organ Size/genetics , Peptide Hormones/pharmacology , Peptide Hormones/physiology , Pituitary Gland/cytology , Pituitary Gland/metabolism , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-fos/metabolism
14.
J Neurosci ; 24(22): 5119-30, 2004 Jun 02.
Article in English | MEDLINE | ID: mdl-15175381

ABSTRACT

G-protein-coupled metabotropic glutamate group I receptors (mGluR1s) mediate synaptic transmission and plasticity in Purkinje cells and, therefore, critically determine cerebellar motor control and learning. Purkinje cells express two members of the G-protein G(q) family, namely G(q) and G11. Although in vitro coexpression of mGluR1 with either Galpha11 or Galpha(q) produces equally well functioning signaling cascades, Galpha(q)- and Galpha11-deficient mice exhibit distinct alterations in motor coordination. By using whole-cell recordings and Ca2+ imaging in Purkinje cells, we show that Galpha(q) is required for mGluR-dependent synaptic transmission and for long-term depression (LTD). Galpha11 has no detectable contribution for synaptic transmission but also contributes to LTD. Quantitative single-cell RT-PCR analyses in Purkinje cells demonstrate a more than 10-fold stronger expression of Galpha(q) versus Galpha11. Our findings suggest an expression level-dependent action of Galpha(q) and Galpha11 for Purkinje cell signaling and assign specific roles of these two G(q) isoforms for motor coordination.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Purkinje Cells/metabolism , Animals , Behavior, Animal/physiology , COS Cells , Calcium/metabolism , Calcium Signaling/genetics , Cerebellum/cytology , Cerebellum/metabolism , Chlorocebus aethiops , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Fluorescent Dyes , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Long-Term Synaptic Depression/genetics , Long-Term Synaptic Depression/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Neuronal Plasticity/physiology , Patch-Clamp Techniques , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Subunits/physiology , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology
15.
Handb Exp Pharmacol ; (165): 437-71, 2004.
Article in English | MEDLINE | ID: mdl-20455101

ABSTRACT

At the injured vessel wall, blood platelets become activated and adhere to the subendothelial surface as well as to each other. These cellular adhesion processes are required for primary hemostasis, but can also lead to thrombosis. Considerable progress has been made during recent years in understanding the molecular mechanisms underlying platelet activation and adhesion. This knowledge will drive future efforts towards the development of new antiplatelet drugs for the prevention and treatment of cardiovascular diseases.

16.
Eur J Neurosci ; 16(12): 2333-41, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12492428

ABSTRACT

Small GTPases of the Rho-family, like Rho, Rac and Cdc42, are involved in neuronal morphogenesis by regulating growth cone morphology or dendritic spine formation. G-proteins of the G12-family, G12 and G13, couple G-protein-coupled receptors (GPCRs) to the activation of RhoA. Recently, two novel Rho-specific guanine nucleotide exchange factors (RhoGEFs), PDZ-RhoGEF and LARG, have been identified to interact with the activated alpha-subunits of G12/G13 and are thus believed to mediate GPCR-induced Rho activation. Although studies in neuronal cell lines have shown that G12/G13 and PDZ-RhoGEF mediate GPCR-induced neurite retraction, the role, as well as the expression of this signalling pathway, in intact brain has not been adequately studied. In the present study, we have characterized systematically the expression of G(alpha)12, G(alpha)13, PDZ-RhoGEF and LARG in various murine tissues as well as their subcellular localization in the central and peripheral nervous systems. By performing immunohistochemistry, using polyclonal antibodies raised against the above proteins, we observed that G(alpha)12, G(alpha)13 and their RhoGEF-effectors are distributed widely in the mammalian nervous system. Moreover, these proteins localize to distinct morphological compartments within neurons. While LARG and G(alpha)12 were mainly found in somata of the neurons, PDZ-RhoGEF and G(alpha)13 were predominantly localized in the neuropil of central neurons. Interestingly, PDZ-RhoGEF is a neural-specific protein, whereas LARG is nearly ubiqoutous. Our data provide evidence that the G12/13-RhoGEF-mediated pathway is present throughout the adult brain and may be involved in regulation of neuronal morphogenesis and function via GPCRs.


Subject(s)
DNA-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Nervous System/metabolism , Neurons/metabolism , Receptors, Glutamate/metabolism , Signal Transduction/physiology , rho GTP-Binding Proteins/metabolism , Animals , Antibody Specificity/immunology , Brain/cytology , Brain/growth & development , Brain/metabolism , DNA, Complementary , GTP-Binding Protein alpha Subunits, G12-G13 , Ganglia, Spinal/cytology , Ganglia, Spinal/growth & development , Ganglia, Spinal/metabolism , Gene Expression Regulation/physiology , Immunohistochemistry , Mice , Nervous System/cytology , Nervous System/growth & development , Neurons/cytology , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Nociceptors/cytology , Nociceptors/metabolism , Rho Guanine Nucleotide Exchange Factors , Spinal Cord/cytology , Spinal Cord/growth & development , Spinal Cord/metabolism
17.
Nat Med ; 7(11): 1236-40, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11689889

ABSTRACT

Myocardial hypertrophy is an adaptational response of the heart to increased work load, but it is also associated with a high risk of cardiac mortality due to its established role in the development of cardiac failure, one of the leading causes of death in developed countries. Multiple growth factors and various downstream signaling pathways involving, for example, ras, gp-130 (ref. 4), JNK/p38 (refs. 5,6) and calcineurin/NFAT/CaM-kinase have been implicated in the hypertrophic response. However, there is evidence that the initial phase in the development of myocardial hypertrophy involves the formation of cardiac para- and/or autocrine factors like endothelin-1, norepinephrine or angiotensin II (refs. 7,8), the receptors of which are coupled to G-proteins of the Gq/11-, G12/13- and Gi/o-families. Cardiomyocyte-specific transgenic overexpression of alpha1-adrenergic or angiotensin (AT1)-receptors as well as of the Gq alpha-subunit, Galphaq, results in myocardial hypertrophy. These data demonstrate that chronic activation of the Gq/G11-family is sufficient to induce myocardial hypertrophy. In order to test whether Gq/G11 mediate the physiological hypertrophy response to pressure overload, we generated a mouse line lacking both Galphaq and Galpha11 in cardiomyocytes. These mice showed no detectable ventricular hypertrophy in response to pressure-overload induced by aortic constriction. The complete lack of a hypertrophic response proves that the Gq/G11-mediated pathway is essential for cardiac hypertrophy induced by pressure overload and makes this signaling process an interesting target for interventions to prevent myocardial hypertrophy.


Subject(s)
Cardiomyopathy, Hypertrophic/prevention & control , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Animals , Base Sequence , Blood Pressure , Cardiomyopathy, Hypertrophic/etiology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , DNA, Complementary/genetics , GTP-Binding Protein alpha Subunits, Gq-G11 , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/physiology , Male , Mice , Mice, Knockout , Mice, Mutant Strains
18.
Z Kardiol ; 90(9): 601-6, 2001 Sep.
Article in German | MEDLINE | ID: mdl-11677795

ABSTRACT

Transmembrane and intracellular signalling pathways are at the center of many cellular regulatory processes. One of the most ubiquitous transmembrane signalling mechanisms involves heterotrimeric guaninnucleotide binding proteins (G-proteins). Members of the Gq/11-family of G-proteins couple receptors to beta-isoforms of phospholipase C. During recent years the establishment of various transgenic animal models has provided new insights into the physiological and pathological role of this signalling pathway. This overview summarizes recent data on the role of Gq/11-mediated signalling processes in platelet activation and myocardial hypertrophy. The increasing knowledge on the molecular mechanisms underlying these processes may lead to new preventive and therapeutic options for the management of coronary artery disease or heart failure.


Subject(s)
Cardiomyopathy, Hypertrophic/physiopathology , Heterotrimeric GTP-Binding Proteins/physiology , Nuclear Proteins/physiology , Platelet Activation/physiology , Signal Transduction/physiology , Animals , Animals, Genetically Modified , Cardiomyopathy, Hypertrophic/genetics , GTP-Binding Protein alpha Subunits, Gq-G11 , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Nuclear Proteins/genetics , Platelet Activation/genetics , Protein Serine-Threonine Kinases , Signal Transduction/genetics
19.
Naunyn Schmiedebergs Arch Pharmacol ; 364(2): 140-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11534853

ABSTRACT

The mammalian G proteins G15 and G16 couple a wide variety of receptors to phospholipase C (PLC) in co-transfected systems, and it has been suggested that they can be used as tools in agonist-screening systems. Using the reversed tetracycline-controlled transactivation system we generated rat pituitary GH3 cell clones that expressed Galphal5 and Galpha16 conditionally to study the coupling of endogenous receptors to both G proteins. In cells expressing moderate levels of Galpha15, activation of various endogenous receptors increased inositol phosphate production, whereas conditional expression of Galpha16 had no significant effect on agonist-dependent PLC activity. Activation of PLC through Galpha15 in response to carbachol did not increase cytosolic [Ca2+] ([Ca2+]i) but stimulated protein kinase C. While carbachol decreased the secretory activity in non-induced GH3 cells, it increased secretion in cells expressing Galpha15. Our data demonstrate that Galpha15 has a higher functional promiscuity than Galpha16 when studied in a system that preserves physiological G protein and receptor levels. In addition, Galpha15-mediated coupling of a receptor to PLC can change the cellular response to receptor agonists, indicating that downstream cellular functions can be used to detect receptor activation in screening systems employing a promiscuous G protein.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Cell Line , Dose-Response Relationship, Drug , Doxycycline/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11 , Heterotrimeric GTP-Binding Proteins/genetics , Inositol Phosphates/metabolism , Plasmids/biosynthesis , Prolactin/metabolism , Protein Kinase C/metabolism , Rats , Receptors, Cell Surface/genetics
20.
J Biol Chem ; 276(51): 47906-13, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11560922

ABSTRACT

Platelets were used to study the activation of Rho and Rac through G-protein-coupled receptors and its regulation by cyclic nucleotides. The thromboxane A(2) (TXA(2)) mimetic rapidly activated both small GTPases independently of integrin alpha(IIb)beta(3) activation., which leads to the activation of G(12)/G(13) and G(q) did not induce Rac activation in G alpha(q)-deficient platelets but was able to activate Rho, to stimulate actin polymerization and phosphatidylinositol 4,5-bisphosphate formation, and to induce shape change. Rac activation by in wild-type platelets could be blocked by chelation of intracellular Ca(2+) and was partially sensitive to apyrase and AR-C69931MX, an antagonist of the G(i)-coupled ADP receptor. Cyclic AMP, which completely blocks platelet function, inhibited the -induced activation of G(q) and G(12)/G(13) as well as of Rac and Rho. In contrast, cGMP, which has no effect on platelet shape change blocked only activation of G(q) and Rac. These data demonstrate that Rho and Rac are differentially regulated through heterotrimeric G-proteins. The G(12)/G(13)-mediated Rho activation is involved in the shape change response, whereas Rac is activated through G(q) and is not required for shape change. Cyclic AMP and cGMP differentially interfere with -induced Rho and Rac activation at least in part by selective effects on the regulation of individual G-proteins through the TXA(2) receptor.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Monophosphate/pharmacology , Animals , Apyrase/pharmacology , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning
SELECTION OF CITATIONS
SEARCH DETAIL
...